Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kuan Chieh Wang is active.

Publication


Featured researches published by Kuan Chieh Wang.


Drug Metabolism and Disposition | 2007

Characterization and Comparative Studies of Zebrafish and Human Recombinant Dihydrofolate Reductases—Inhibition by Folic Acid and Polyphenols

Tseng Ting Kao; Kuan Chieh Wang; Wen Ni Chang; Chia-Ying Lin; Bing Hung Chen; Hua-Lin Wu; Guey-Yueh Shi; Jen Ning Tsai; Tzu Fun Fu

Dihydrofolate reductase (DHFR) catalyzes folic acid reduction and recycles dihydrofolate generated during dTMP biosynthesis to tetrahydrofolate. DHFR is the main target of methotrexate, the most widely used agent for antifolate therapy. Nevertheless, the emergence of methotrexate-resistance has greatly impeded the curative potential of this drug. Therefore, drugs with improved efficacy are still in demand, as well as an efficient in vitro assay system and animal model for antifolate drug discovery. The aim of this study is to evaluate the suitability of using zebrafish DHFR as an alternative assay system for antifolate drug discovery. The cDNAs encoding zebrafish and human DHFR were cloned, overexpressed, and purified. Similar structural and kinetic properties were revealed between zebrafish and human recombinant DHFRs. The susceptibilities of both enzymes to known DHFR inhibitors, including methotrexate and trimethoprim, and compounds with antifolate potential, such as polyphenols, are also comparable. In addition, the DHFR-mediated dihydrofolate reduction was significantly inhibited by its own substrate folic acid. An unexpected tissue-specific distribution of DHFR was observed with the highest level present in ova and brains of zebrafish. DHFR is also abundant in zebrafish embryos of early stages and decreased abruptly after 3 days postfertilization. The substantial resemblance between zebrafish and human DHFRs, as demonstrated in this study, provides compelling evidence supporting the use of zebrafish DHFR as an in vitro assay system for folate-related studies and drug discovery.


PLOS ONE | 2012

The Epidermal Growth Factor-like Domain of CD93 Is a Potent Angiogenic Factor

Yuan Chung Kao; Shinn-Jong Jiang; Wen An Pan; Kuan Chieh Wang; Po Ku Chen; Hsi Ju Wei; Wei Sheng Chen; Bi Ing Chang; Guey-Yueh Shi; Hua-Lin Wu

Human CD93, an epidermal growth factor (EGF)-like domain containing transmembrane protein, is predominantly expressed in the vascular endothelium. Studies have shown that AA4, the homolog of CD93 in mice, may mediate cell migration and angiogenesis in endothelial cells. Soluble CD93 has been detected in the plasma of healthy individuals. However, the role of soluble CD93 in the endothelium remains unclear. Recombinant soluble CD93 proteins with EGF-like domains (rCD93D123, with domains 1, 2, and 3; and rCD93D23, with domains 2 and 3) were generated to determine their functions in angiogenesis. We found that rCD93D23 was more potent than rCD93D123 in stimulating the proliferation and migration of human umbilical vein endothelial cells (HUVECs). Production of matrix-metalloproteinase 2 increased after the HUVECs were treated with rCD93D23. Further, in a tube formation assay, rCD93D23 induced cell differentiation of HUVECs through phosphoinositide 3-kinase/Akt/endothelial nitric oxide synthase and extracellular signal-regulated kinases-1/2 signaling. Moreover, rCD93D23 promoted blood vessel formation in a Matrigel-plug assay and an oxygen-induced retinopathy model in vivo. Our findings suggest that the soluble EGF-like domain containing CD93 protein is a novel angiogenic factor acting on the endothelium.


Journal of Investigative Dermatology | 2013

Thrombomodulin Regulates Keratinocyte Differentiation and Promotes Wound Healing

Tsung Lin Cheng; Yu Ting Wu; Chao Han Lai; Yuan Chung Kao; Cheng Hsiang Kuo; Shu Lin Liu; Yun Yan Hsu; Po Ku Chen; Chia Fong Cho; Kuan Chieh Wang; Wei Ling Lin; Bi Ing Chang; Chun Ming Chen; Hartmut Weiler; Guey-Yueh Shi; Hua-Lin Wu

The membrane glycoprotein thrombomodulin (TM) has been implicated in keratinocyte differentiation and wound healing, but its specific function remains undetermined. The epidermis-specific TM knockout mice were generated to investigate the function of TM in these biological processes. Primary cultured keratinocytes obtained from TM(lox/lox); K5-Cre mice, in which TM expression was abrogated, underwent abnormal differentiation in response to calcium induction. Poor epidermal differentiation, as evidenced by downregulation of the terminal differentiation markers loricrin and filaggrin, was observed in TM(lox/lox); K5-Cre mice. Silencing TM expression in human epithelial cells impaired calcium-induced extracellular signal-regulated kinase pathway activation and subsequent keratinocyte differentiation. Compared with wild-type mice, the cell spreading area and wound closure rate were lower in keratinocytes from TM(lox/lox); K5-Cre mice. In addition, the lower density of neovascularization and smaller area of hyperproliferative epithelium contributed to slower wound healing in TM(lox/lox); K5-Cre mice than in wild-type mice. Local administration of recombinant TM (rTM) accelerated healing rates in the TM-null skin. These data suggest that TM has a critical role in skin differentiation and wound healing. Furthermore, rTM may hold therapeutic potential for the treatment of nonhealing chronic wounds.


Journal of Thrombosis and Haemostasis | 2010

Human plasminogen kringle 1–5 reduces atherosclerosis and neointima formation in mice by suppressing the inflammatory signaling pathway

Po Chiao Chang; Hua-Lin Wu; Huey-Juan Lin; Kuan Chieh Wang; Guey-Yueh Shi

Summary.  Background: Activation of vascular endothelial cells plays an important role in atherogenesis and plaque instability. Recent research has demonstrated that late‐stage inhibition of plaque angiogenesis by angiostatin (kringle 1–4) reduces macrophage accumulation and slows the progression of advanced atherosclerosis. Kringle 1–5 (K1–5) is a variant of angiostatin that contains the first five kringle domains of plasminogen. Objective: To investigate whether K1–5 has an inhibitory effect on early‐stage atherosclerosis, using the apolipoprotein E (ApoE)‐deficient mouse model and a carotid artery ligation model. Methods: ApoE‐deficient mice received K1–5 treatment for 4 weeks, and the severity of aortic atherosclerosis was measured. In the ligation model, the left common carotid arteries of C57BL/6 mice were ligated near the carotid bifurcation, and the mice received K1–5 for 4 weeks. Human umbilical vein endothelial cells were pretreated with K1–5 before tumor necrosis factor‐α (TNF‐α) treatment to explore the anti‐inflammatory effect of K1–5. Results: The areas of the lesion in the aortas of ApoE‐deficient mice that received K1–5 treatment were notably decreased, and the formation of carotid neointima in the C57BL/6 mice was decreased by treatment with K1–5. Expression of TNF‐α‐induced intercellular adhesion molecule‐1 and vascular cell adhesion molecule‐1 was inhibited by K1–5 treatment, possibly via downregulation of translocation of nuclear factor‐κB and expression of reactive oxygen species. Conclusions: K1–5 reduced atherosclerosis and neointima formation in mice, possibly through inhibition of intercellular adhesion molecule‐1 and vascular cell adhesion molecule‐1 expression in endothelial cells.


PLOS ONE | 2016

Toll-Like Receptor 4 Is Essential in the Development of Abdominal Aortic Aneurysm.

Chao Han Lai; Kuan Chieh Wang; Fang Tzu Lee; Hung Wen Tsai; Chih Yuan Ma; Tsung Lin Cheng; Bi Ing Chang; Yu Jen Yang; Guey-Yueh Shi; Hua-Lin Wu

Toll-like receptor (TLR) family plays a key role in innate immunity and various inflammatory responses. TLR4, one of the well-characterized pattern-recognition receptors, can be activated by endogenous damage-associated molecular pattern molecules such as high mobility group box 1 (HMGB1) to sustain sterile inflammation. Evidence suggested that blockade of TLR4 signaling may confer protection against abdominal aortic aneurysm (AAA). Herein we aimed to obtain further insight into the mechanism by which TLR4 might promote aneurysm formation. Characterization of the CaCl2-induced AAA model in mice revealed that upregulation of TLR4 expression, localized predominantly to vascular smooth muscle cells (VSMCs), was followed by a late decline during a 28-day period of AAA development. In vitro, TLR4 expression was increased in VSMCs treated with HMGB1. Knockdown of TLR4 by siRNA attenuated HMGB1-enhanced production of proinflammatory cytokines, specifically interleukin-6 and monocyte chemoattractant protein-1 (MCP-1), and matrix-degrading matrix metalloproteinase (MMP)-2 from VSMCs. In vivo, two different strains of TLR4-deficient (C57BL/10ScNJ and C3H/HeJ) mice were resistant to CaCl2-induced AAA formation compared to their respective controls (C57BL/10ScSnJ and C3H/HeN). Knockout of TLR4 reduced interleukin-6 and MCP-1 levels and HMGB1 expression, attenuated macrophage accumulation, and eventually suppressed MMP production, elastin destruction and VSMC loss. Finally, human AAA exhibited higher TLR4 expression that was localized to VSMCs. These data suggest that TLR4 signaling contributes to AAA formation by promoting a proinflammatory status of VSMCs and by inducing proteinase release from VSMCs during aneurysm initiation and development.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Membrane-Bound Thrombomodulin Regulates Macrophage Inflammation in Abdominal Aortic Aneurysm

Kuan Chieh Wang; Yi-Heng Li; Guey-Yueh Shi; Hung Wen Tsai; Chawn Yau Luo; Min Hua Cheng; Chih Yuan Ma; Yun Yan Hsu; Tsung Lin Cheng; Bi Ing Chang; Chao Han Lai; Hua-Lin Wu

Objective—Thrombomodulin (TM), a glycoprotein constitutively expressed in the endothelium, is well known for its anticoagulant and anti-inflammatory properties. Paradoxically, we recently found that monocytic membrane-bound TM (ie, endogenous TM expression in monocytes) triggers lipopolysaccharide- and gram-negative bacteria–induced inflammatory responses. However, the significance of membrane-bound TM in chronic sterile vascular inflammation and the development of abdominal aortic aneurysm (AAA) remains undetermined. Approach and Results—Implicating a potential role for membrane-bound TM in AAA, we found that TM signals were predominantly localized to macrophages and vascular smooth muscle cells in human aneurysm specimens. Characterization of the CaCl2-induced AAA in mice revealed that during aneurysm development, TM expression was mainly localized in infiltrating macrophages and vascular smooth muscle cells. To investigate the function of membrane-bound TM in vivo, transgenic mice with myeloid- (LysMcre/TMflox/flox) and vascular smooth muscle cell–specific (SM22-cretg/TMflox/flox) TM ablation and their respective wild-type controls (TMflox/flox and SM22-cretg/TM+/+) were generated. In the mouse CaCl2-induced AAA model, deficiency of myeloid TM, but not vascular smooth muscle cell TM, inhibited macrophage accumulation, attenuated proinflammatory cytokine and matrix metalloproteinase-9 production, and finally mitigated elastin destruction and aortic dilatation. In vitro TM-deficient monocytes/macrophages, versus TM wild-type counterparts, exhibited attenuation of proinflammatory mediator expression, adhesion to endothelial cells, and generation of reactive oxygen species. Consistently, myeloid TM–deficient hyperlipidemic mice (ApoE−/−/LysMcre/TMflox/flox) were resistant to AAA formation induced by angiotensin II infusion, along with reduced macrophage infiltration, suppressed matrix metalloproteinase activities, and diminished oxidative stress. Conclusions—Membrane-bound TM in macrophages plays an essential role in the development of AAA by enhancing proinflammatory mediator elaboration, macrophage recruitment, and oxidative stress.


Journal of Investigative Dermatology | 2015

Thrombomodulin promotes diabetic wound healing by regulating toll-like receptor 4 expression

Tsung Lin Cheng; Chao Han Lai; Po Ku Chen; Chia Fong Cho; Yun Yan Hsu; Kuan Chieh Wang; Wei Ling Lin; Bi Ing Chang; Shi Kai Liu; Yu Ting Wu; Chao Kai Hsu; Guey-Yueh Shi; Hua-Lin Wu

Keratinocyte-expressed thrombomodulin (TM) and the released soluble TM (sTM) have been demonstrated to promote wound healing. However, the effects of high glucose on TM expression in keratinocytes and the role of TM in diabetic ulcer remain unclear. In this study, we demonstrated that expressions of TM and Toll-like receptor 4 (TLR4) were both downregulated in high-glucose cultured human keratinocytes and in skin keratinocytes of diabetic patients. In addition, the wound-triggered upregulation of TM and sTM production was abolished in both high-glucose cultured human keratinocytes and streptozotocin-induced diabetic mouse skin. Furthermore, supplementation of recombinant sTM could increase TLR4 expression and promote cutaneous wound healing in both high-glucose cultured human keratinocytes and diabetic mice. However, in Tlr4-deleted mice, which exhibited delayed wound healing, the therapeutic benefit of recombinant sTM was abrogated. Moreover, our results showed that tumor necrosis factor-α (TNF-α) expression in keratinocytes was dose-dependently upregulated by glucose, and TNF-α treatment downregulated the expression of TM and TLR4. Taken together, high-glucose environment reduces the expression of TM and TLR4 in keratinocytes possibly through the action of TNF-α, and recombinant sTM can increase the TLR4 expression and promote wound healing under diabetic condition.


Oncotarget | 2016

Thrombomodulin promotes focal adhesion kinase activation and contributes to angiogenesis by binding to fibronectin

Yun-Yan Hsu; Guey-Yueh Shi; Kuan Chieh Wang; Chih-Yuan Ma; Tsung-Lin Cheng; Hua-Lin Wu

Angiogenesis promotes tumor growth and metastasis. Cell adhesion molecules interact with the extracellular matrix (ECM) and increase cell adhesion and migration during angiogenesis. Thrombomodulin (TM) is a cell surface transmembrane glycoprotein expressed in endothelial cells. However, the function and significance of TM in cell-matrix interactions and angiogenesis remain unclear. Here, we first demonstrated that recombinant lectin-like domain of TM interacts with an ECM protein, fibronectin, and identified the N-terminal 70-kDa domain of fibronectin as the TM-binding site. Exogenous expression of TM in TM-deficient A2058 melanoma cells enhanced cell adhesion and migration on fibronectin and invasion on Matrigel. In addition, TM increased focal adhesion kinase (FAK) phosphorylation and matrix metalloproteinase-9 production. In mice bearing subcutaneous B16F10 melanoma tumors, immunofluorescence analysis indicated that TM was highly expressed and co-localized with fibronectin on the tumor vasculature. The interaction between TM and fibronectin in tumor blood vessels was also validated by the proximity ligation assay. In human umbilical vein endothelial cells, up-regulation of TM by vascular endothelial growth factor (VEGF), a tumor angiogenic factor, promoted cell adhesion and tube formation, whereas TM knockdown by RNA interference attenuated VEGF-induced cell adhesion and tube formation. In summary, TM promotes angiogenesis by enhancing cell adhesion, migration, and FAK activation through interaction with fibronectin. TM may represent a novel target for inhibiting tumor angiogenesis.


Hypertension | 2016

Peroxisome Proliferator–Activated Receptor γ Level Contributes to Structural Integrity and Component Production of Elastic Fibers in the Aorta

Haw Chih Tai; Pei-Jane Tsai; Ju Yi Chen; Chao Han Lai; Kuan Chieh Wang; Shih Hua Teng; Shih Chieh Lin; Alice Y.W. Chang; Meei Jyh Jiang; Yi-Heng Li; Hua-Lin Wu; Nobuyo Maeda; Yau Sheng Tsai

Loss of integrity and massive disruption of elastic fibers are key features of abdominal aortic aneurysm (AAA). Peroxisome proliferator–activated receptor &ggr; (PPAR&ggr;) has been shown to attenuate AAA through inhibition of inflammation and proteolytic degradation. However, its involvement in elastogenesis during AAA remains unclear. PPAR&ggr; was highly expressed in human AAA within all vascular cells, including inflammatory cells and fibroblasts. In the aortas of transgenic mice expressing PPAR&ggr; at 25% normal levels (PpargC/− mice), we observed the fragmentation of elastic fibers and reduced expression of vital elastic fiber components of elastin and fibulin-5. These were not observed in mice with 50% normal PPAR&ggr; expression (Pparg+/− mice). Infusion of a moderate dose of angiotensin II (500 ng/kg per minute) did not induce AAA but Pparg+/− aorta developed flattened elastic lamellae, whereas PpargC/− aorta showed severe destruction of elastic fibers. After infusion of angiotensin II at 1000 ng/kg per minute, 73% of PpargC/− mice developed atypical suprarenal aortic aneurysms: superior mesenteric arteries were dilated with extensive collagen deposition in adventitia and infiltrations of inflammatory cells. Although matrix metalloproteinase inhibition by doxycycline somewhat attenuated the dilation of aneurysm, it did not reduce the incidence nor elastic lamella deterioration in angiotensin II–infused PpargC/− mice. Furthermore, PPAR&ggr; antagonism downregulated elastin and fibulin-5 in fibroblasts, but not in vascular smooth muscle cells. Chromatin immunoprecipitation assay demonstrated PPAR&ggr; binding in the genomic sequence of fibulin-5 in fibroblasts. Our results underscore the importance of PPAR&ggr; in AAA development though orchestrating proper elastogenesis and preserving elastic fiber integrity.


Scientific Reports | 2017

Recombinant Thrombomodulin Exerts Anti-autophagic Action in Endothelial Cells and Provides Anti-atherosclerosis Effect in Apolipoprotein E Deficient Mice

Po Sheng Chen; Kuan Chieh Wang; Ting-Hsing Chao; Hsing Chun Chung; Shi Ya Tseng; Chawn Yau Luo; Guey-Yueh Shi; Hua-Lin Wu; Yi-Heng Li

Stress-induced alteration in endothelial cells (ECs) integrity precedes the development of atherosclerosis. Previous studies showed that the soluble recombinant thrombomodulin (rTM) not only increases ECs proliferation but also exerts anti-apoptotic activity in ECs. However, the functional significance of soluble rTM on autophagy-related apoptosis in ECs is still undetermined. Implicating a cytoprotective role for rTM in persistent serum starvation (SS)-induced autophagy in cultured ECs, we found that treatment of rTM decreased the expression of SS-induced autophagy-related proteins, ATG5 and LC3, and the formation of autophagosomes through activation of AKT/mTOR pathway. In addition, treatment of rTM decreased SS-induced EC apoptosis, but this effect of rTM could not be recapitulated by co-treatment with a potent autophagy inducer, rapamycin and in ECs with ATG5 knockdown. In human atherosclerosis specimens, expression of autophagy markers, ATG13 and LC3, were more abundant in aortic intimal ECs with severe atherosclerosis than those without atherosclerosis. Moreover, compared to saline treatment group, administration of rTM reduced LC3 and ATG13 expression, intimal EC apoptosis, and atherosclerotic lesion severity in the aorta of apolipoprotein E deficient mice. In conclusion, treatment with rTM suppressed stress-induced autophagy overactivation in ECs, provided ECs protective effects, and decreased atherosclerosis in apolipoprotein E deficient mice.

Collaboration


Dive into the Kuan Chieh Wang's collaboration.

Top Co-Authors

Avatar

Hua-Lin Wu

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Guey-Yueh Shi

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Chao Han Lai

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Bi Ing Chang

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Tsung Lin Cheng

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Yi-Heng Li

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Cheng Hsiang Kuo

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Chih Yuan Ma

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Fang Tzu Lee

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Po Ku Chen

National Cheng Kung University

View shared research outputs
Researchain Logo
Decentralizing Knowledge